1
|
Shaygani H, Mofrad YM, Demneh SMR, Hafezi S, Almasi-Jaf A, Shamloo A. Cartilage and bone injectable hydrogels: A review of injectability methods and treatment strategies for repair in tissue engineering. Int J Biol Macromol 2024; 282:136689. [PMID: 39447779 DOI: 10.1016/j.ijbiomac.2024.136689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Cartilage and bone are crucial tissues causing disability in the elderly population, often requiring prolonged treatment and surgical intervention due to limited regenerative capacity. Injectable hydrogels that closely mimic the extracellular matrix (ECM) of native hard tissue have attracted attention due to their minimally invasive application and ability to conform to irregular defect sites. These hydrogels facilitate key biological processes such as cell migration, chondrogenesis in cartilage repair, osteoinduction, angiogenesis, osteoconduction, and mineralization in bone repair. This review analyzes in-vitro and in-vivo biomedical databases over the past decade to identify advancements in hydrogel formulations, crosslinking mechanisms, and biomaterial selection for cartilage and bone tissue engineering. The review emphasizes the effectiveness of injectable hydrogels as carriers for cells, growth factors, and drugs, offering additional therapeutic benefits. The relevance of these findings is discussed in the context of their potential to serve as a robust alternative to current surgical and non-surgical treatments. This review also examines the advantages of injectable hydrogels, such as ease of administration, reduced patient recovery time, and enhanced bioactivity, thereby emphasizing their potential in clinical applications for cartilage and bone regeneration with emphasis on addressing the shortcomings of current treatments.
Collapse
Affiliation(s)
- Hossein Shaygani
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Yasaman Mozhdehbakhsh Mofrad
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran; School of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Mohammadhossein Rezaei Demneh
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Shayesteh Hafezi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Aram Almasi-Jaf
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Amir Shamloo
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
2
|
Sun X, Wang N, Jiang H, Liu Q, Xiao C, Xu J, Wu Y, Mei J, Wu S, Lin Z. Insulin-transferrin-selenium promote formation of tissue-engineered vascular grafts in early stage of culture. Prep Biochem Biotechnol 2024; 54:1186-1195. [PMID: 38546975 DOI: 10.1080/10826068.2024.2333468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
To create tissue-engineered vascular grafts (TEVGs) in vitro, vascular smooth muscle cells (VSMCs) must function effectively and produce sufficient extracellular matrix (ECM) in a three-dimensional space. In this study, we investigated whether the addition of insulin-transferrin-selenium (ITS), a medium supplement, could enhance TEVG formation. PGA fabric was used as the scaffold, and 1% ITS was added to the medium. After two weeks, the tissues were examined using electron microscopy and staining. The ITS group exhibited a denser structure and increased collagen production. VSMCs were cultured in two dimensions with ITS and assessed for collagen production, cell growth, and glucose metabolism. The results showed that ITS supplementation increased collagen production, cell growth, glucose utilization, lactate production, and ATP levels. Furthermore, reducing the amount of fetal bovine serum (FBS) in the medium did not affect the TEVGs or VSMCs when ITS was present. In conclusion, ITS improves TEVG construction by promoting VSMCs growth and reducing the need for FBS.
Collapse
MESH Headings
- Tissue Engineering/methods
- Insulin/metabolism
- Animals
- Blood Vessel Prosthesis
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Selenium/pharmacology
- Selenium/chemistry
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Cells, Cultured
- Cell Proliferation/drug effects
- Rats
- Tissue Scaffolds/chemistry
- Collagen/metabolism
- Glucose/metabolism
Collapse
Affiliation(s)
- Xuheng Sun
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| | - Nannan Wang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
| | - Hongjing Jiang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| | - Qing Liu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| | - Cong Xiao
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| | - Jianyi Xu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| | - Yindi Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| | - Jingyi Mei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
| | - Shuting Wu
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Province, P.R. China
| | - Zhanyi Lin
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, P.R. China
- JIHUA Laboratory, Foshan City, Guangdong Province, P.R. China
| |
Collapse
|
3
|
Tian C, Li K, Chu F, Wei Q, Xu S, Qiang L, Gou X. Preparation and performance study of in situ mineralized bone tissue engineering scaffolds. RSC Adv 2024; 14:22420-22433. [PMID: 39010908 PMCID: PMC11248912 DOI: 10.1039/d4ra04047c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Traditional bone tissue engineering techniques require the extraction and proliferation of seed cells, followed by prolonged in vitro culture to form bone tissue constructs. In contrast, in situ mineralization bone tissue engineering utilizes alkaline phosphatase within the body's microenvironment to induce scaffold mineralization. This approach promotes further proliferation and differentiation of osteoblasts and the formation of bone tissue constructs, thereby simplifying the traditional bone tissue engineering process. This study uses electrospinning technology to prepare a novel biologically active scaffold for bone tissue engineering using poly(lactic-co-glycolic acid) (PLGA) and calcium glycerophosphate. The morphology and composition of the scaffolds were characterized using SEM, EDS, and XRD, revealing well-defined fibrous structures and the successful incorporation of calcium glycerophosphate into the PLGA fibers. In vitro simulation of the bone microenvironment using alkaline phosphatase effectively catalyzed the in situ mineralization of calcium glycerophosphate within the scaffold. SEM observations showed substantial mineral aggregation on the surface of the fibrous membranes, and XRD characterization confirmed that the diffraction peaks of the minerals correspond to hydroxyapatite. The cytotoxicity, cell proliferation, and osteogenic differentiation assessments on MC3T3-E1 pre-osteoblasts cultured on the prepared scaffolds indicate that the scaffolds are non-toxic to cells and possess good osteogenic differentiation ability, enabling in situ mineralization. This suggests that the scaffolds have broad prospects for application in bone defect repair.
Collapse
Affiliation(s)
- Chunyan Tian
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
| | - Kun Li
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
| | - Fuhuan Chu
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
| | - Qiujiang Wei
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
| | - Shiqi Xu
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
- Hebei International Research Center for Medical-Engineering, Chengde Medical University Chengde 067000 Hebei China
- Chengde Medical Additive Manufacturing Technology Innovation Center, Chengde Medical University Chengde 067000 Hebei China
| | - Linhui Qiang
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
- Hebei International Research Center for Medical-Engineering, Chengde Medical University Chengde 067000 Hebei China
- Chengde Medical Additive Manufacturing Technology Innovation Center, Chengde Medical University Chengde 067000 Hebei China
| | - Xinrui Gou
- Department of Biomedical Engineering, Chengde Medical University Chengde 067000 Hebei China +86-13343396119 +86-17774937339
| |
Collapse
|
4
|
Feng K, Wang F, Chen H, Zhang R, Liu J, Li X, Xie X, Kang Q. Cartilage progenitor cells derived extracellular vesicles-based cell-free strategy for osteoarthritis treatment by efficient inflammation inhibition and extracellular matrix homeostasis restoration. J Nanobiotechnology 2024; 22:345. [PMID: 38890638 PMCID: PMC11186174 DOI: 10.1186/s12951-024-02632-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease which currently lacks of effective agents. It is therefore urgent and necessary to seek an effective approach that can inhibit inflammation and promote cartilage matrix homeostasis. Cartilage progenitor cells (CPCs) are identified as a cell population of superficial zone in articular cartilage which possess strong migration ability, proliferative capacity, and chondrogenic potential. Recently, the application of CPCs may represent a novel cell therapy strategy for OA treatment. There is growing evidence that extracellular vesicles (EVs) are primary mediators of the benefits of stem cell-based therapy. In this study, we explored the protective effects of CPCs-derived EVs (CPCs-EVs) on IL-1β-induced chondrocytes. We found CPCs-EVs exhibited chondro-protective effects in vitro. Furthermore, our study demonstrated that CPCs-EVs promoted matrix anabolism and inhibited inflammatory response at least partially via blocking STAT3 activation. In addition, liquid chromatography-tandem mass spectrometry analysis identified 991 proteins encapsulated in CPCs-EVs. By bioinformatics analysis, we showed that STAT3 regulatory proteins were enriched in CPCs-EVs and could be transported to chondrocytes. To promoting the protective function of CPCs-EVs in vivo, CPCs-EVs were modified with cationic peptide ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) for surface charge reverse. In posttraumatic OA mice, our results showed PPD modified CPCs-EVs (PPD-EVs) effectively inhibited extracellular matrix catabolism and attenuated cartilage degeneration. Moreover, PPD-EVs down-regulated inflammatory factors expressions and reduced OA-related pain in OA mice. In ex-vivo cultured OA cartilage explants, PPD-EVs successfully promoted matrix anabolism and inhibited inflammation. Collectively, CPCs-EVs-based cell-free therapy is a promising strategy for OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongfang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Rui Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiashuo Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaodong Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuetao Xie
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
5
|
Chen L, Yang J, Cai Z, Huang Y, Xiao P, Wang J, Wang F, Huang W, Cui W, Hu N. Electroactive Biomaterials Regulate the Electrophysiological Microenvironment to Promote Bone and Cartilage Tissue Regeneration. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202314079] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Indexed: 01/06/2025]
Abstract
AbstractThe incidence of large bone and articular cartilage defects caused by traumatic injury is increasing worldwide; the tissue regeneration process for these injuries is lengthy due to limited self‐healing ability. Endogenous bioelectrical phenomenon has been well recognized to play an important role in bone and cartilage homeostasis and regeneration. Studies have reported that electrical stimulation (ES) can effectively regulate various biological processes and holds promise as an external intervention to enhance the synthesis of the extracellular matrix, thereby accelerating the process of bone and cartilage regeneration. Hence, electroactive biomaterials have been considered a biomimetic approach to ensure functional recovery by integrating various physiological signals, including electrical, biochemical, and mechanical signals. This review will discuss the role of endogenous bioelectricity in bone and cartilage tissue, as well as the effects of ES on cellular behaviors. Then, recent advances in electroactive materials and their applications in bone and cartilage tissue regeneration are systematically overviewed, with a focus on their advantages and disadvantages as tissue repair materials and performances in the modulation of cell fate. Finally, the significance of mimicking the electrophysiological microenvironment of target tissue is emphasized and future development challenges of electroactive biomaterials for bone and cartilage repair strategies are proposed.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Jianye Yang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Zhengwei Cai
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Yanran Huang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Pengcheng Xiao
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Juan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Fan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Wei Huang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Ning Hu
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| |
Collapse
|
6
|
Nordberg RC, Bielajew BJ, Takahashi T, Dai S, Hu JC, Athanasiou KA. Recent advancements in cartilage tissue engineering innovation and translation. Nat Rev Rheumatol 2024; 20:323-346. [PMID: 38740860 PMCID: PMC11524031 DOI: 10.1038/s41584-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/16/2024]
Abstract
Articular cartilage was expected to be one of the first successfully engineered tissues, but today, cartilage repair products are few and they exhibit considerable limitations. For example, of the cell-based products that are available globally, only one is marketed for non-knee indications, none are indicated for severe osteoarthritis or rheumatoid arthritis, and only one is approved for marketing in the USA. However, advances in cartilage tissue engineering might now finally lead to the development of new cartilage repair products. To understand the potential in this field, it helps to consider the current landscape of tissue-engineered products for articular cartilage repair and particularly cell-based therapies. Advances relating to cell sources, bioactive stimuli and scaffold or scaffold-free approaches should now contribute to progress in therapeutic development. Engineering for an inflammatory environment is required because of the need for implants to withstand immune challenge within joints affected by osteoarthritis or rheumatoid arthritis. Bringing additional cartilage repair products to the market will require an understanding of the translational vector for their commercialization. Advances thus far can facilitate the future translation of engineered cartilage products to benefit the millions of patients who suffer from cartilage injuries and arthritides.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Takumi Takahashi
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Shuyan Dai
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
7
|
Majumder N, Roy C, Doenges L, Martin I, Barbero A, Ghosh S. Covalent Conjugation of Small Molecule Inhibitors and Growth Factors to a Silk Fibroin-Derived Bioink to Develop Phenotypically Stable 3D Bioprinted Cartilage. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9925-9943. [PMID: 38362893 DOI: 10.1021/acsami.3c18903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Implantation of a phenotypically stable cartilage graft could represent a viable approach for repairing osteoarthritic (OA) cartilage lesions. In the present study, we investigated the effects of modulating the bone morphogenetic protein (BMP), transforming growth factor beta (TGFβ), and interleukin-1 (IL-1) signaling cascades in human bone marrow stromal cell (hBMSC)-encapsulated silk fibroin gelatin (SF-G) bioink. The selected small molecules LDN193189, TGFβ3, and IL1 receptor antagonist (IL1Ra) are covalently conjugated to SF-G biomaterial to ensure sustained release, increased bioavailability, and printability, confirmed by ATR-FTIR, release kinetics, and rheological analyses. The 3D bioprinted constructs with chondrogenically differentiated hBMSCs were incubated in an OA-inducing medium for 14 days and assessed through a detailed qPCR, immunofluorescence, and biochemical analyses. Despite substantial heterogeneity in the observations among the donors, the IL1Ra molecule illustrated the maximum efficiency in enhancing the expression of articular cartilage components, reducing the expression of hypertrophic markers (re-validated by the GeneMANIA tool), as well as reducing the production of inflammatory molecules by the hBMSCs. Therefore, this study demonstrated a novel strategy to develop a chemically decorated, printable and biomimetic SF-G bioink to produce hyaline cartilage grafts resistant to acquiring OA traits that can be used for the treatment of degenerated cartilage lesions.
Collapse
Affiliation(s)
- Nilotpal Majumder
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Chandrashish Roy
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Laura Doenges
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
8
|
Jin A, del Valle LJ, Puiggalí J. Copolymers and Blends Based on 3-Hydroxybutyrate and 3-Hydroxyvalerate Units. Int J Mol Sci 2023; 24:17250. [PMID: 38139077 PMCID: PMC10743438 DOI: 10.3390/ijms242417250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
This review presents a comprehensive update of the biopolymer poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV), emphasizing its production, properties, and applications. The overall biosynthesis pathway of PHBV is explored in detail, highlighting recent advances in production techniques. The inherent physicochemical properties of PHBV, along with its degradation behavior, are discussed in detail. This review also explores various blends and composites of PHBV, demonstrating their potential for a range of applications. Finally, the versatility of PHBV-based materials in multiple sectors is examined, emphasizing their increasing importance in the field of biodegradable polymers.
Collapse
Affiliation(s)
- Anyi Jin
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, EEBE, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain; (A.J.); (L.J.d.V.)
- Venvirotech Biotechnology S.L., Santa Perpètua de Mogoda, 08130 Barcelona, Spain
| | - Luis J. del Valle
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, EEBE, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain; (A.J.); (L.J.d.V.)
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Campus Diagonal-Besòs, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Jordi Puiggalí
- Departament d’Enginyeria Química, Universitat Politècnica de Catalunya, EEBE, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain; (A.J.); (L.J.d.V.)
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Campus Diagonal-Besòs, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain
| |
Collapse
|
9
|
Gu Y, Hu Y, Zhang H, Wang S, Xu K, Su J. Single-cell RNA sequencing in osteoarthritis. Cell Prolif 2023; 56:e13517. [PMID: 37317049 PMCID: PMC10693192 DOI: 10.1111/cpr.13517] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/30/2023] [Accepted: 05/26/2023] [Indexed: 06/16/2023] Open
Abstract
Osteoarthritis is a progressive and heterogeneous joint disease with complex pathogenesis. The various phenotypes associated with each patient suggest that better subgrouping of tissues associated with genotypes in different phases of osteoarthritis may provide new insights into the onset and progression of the disease. Recently, single-cell RNA sequencing was used to describe osteoarthritis pathogenesis on a high-resolution view surpassing traditional technologies. Herein, this review summarizes the microstructural changes in articular cartilage, meniscus, synovium and subchondral bone that are mainly due to crosstalk amongst chondrocytes, osteoblasts, fibroblasts and endothelial cells during osteoarthritis progression. Next, we focus on the promising targets discovered by single-cell RNA sequencing and its potential applications in target drugs and tissue engineering. Additionally, the limited amount of research on the evaluation of bone-related biomaterials is reviewed. Based on the pre-clinical findings, we elaborate on the potential clinical values of single-cell RNA sequencing for the therapeutic strategies of osteoarthritis. Finally, a perspective on the future development of patient-centred medicine for osteoarthritis therapy combining other single-cell multi-omics technologies is discussed. This review will provide new insights into osteoarthritis pathogenesis on a cellular level and the field of applications of single-cell RNA sequencing in personalized therapeutics for osteoarthritis in the future.
Collapse
Affiliation(s)
- Yuyuan Gu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- School of MedicineShanghai UniversityShanghaiChina
| | - Yan Hu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
| | - Hao Zhang
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
| | - Sicheng Wang
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Department of OrthopedicsShanghai Zhongye HospitalShanghaiChina
| | - Ke Xu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Wenzhou Institute of Shanghai UniversityWenzhouChina
| | - Jiacan Su
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
| |
Collapse
|
10
|
Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels: An emerging therapeutic strategy for cartilage regeneration. Adv Colloid Interface Sci 2023; 321:103030. [PMID: 37907031 DOI: 10.1016/j.cis.2023.103030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Martyn Snow
- Department of Arthroscopy, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham B31 2AP, United Kingdom; The Robert Jones and Agnes Hunt Hospital, Oswestry, Shropshire SY10 7AG, United Kingdom
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom; Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Pooya Davoodi
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom.
| |
Collapse
|
11
|
Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater 2023; 168:372-387. [PMID: 37481194 DOI: 10.1016/j.actbio.2023.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/26/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Orthopedics, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
12
|
Zhang C, Wang G, An Y. Achieving Nasal Septal Cartilage In Situ Regeneration: Focus on Cartilage Progenitor Cells. Biomolecules 2023; 13:1302. [PMID: 37759702 PMCID: PMC10527213 DOI: 10.3390/biom13091302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
The nasal septal cartilage plays an important role in preventing the collapse of the nasal bones and maintaining the appearance of the nose. In the context of inherent difficulties regarding septal cartilage repair and the shortage of cartilage graft resources for regeneration, tissue engineering, especially the in situ strategy based on scaffolds, has become a new prospect and become one of the most promising approaches. Given that it is difficult for chondrocytes to achieve directional migration and secrete matrix components to participate in tissue repair after cartilage injury, cartilage progenitor cells (CPCs), with great migratory ability and stem cell characteristics, have caught the attention of researchers and brought hope for nasal septal cartilage in situ regeneration. In this review, we first summarized the distribution, characteristics, isolation, and culture methods of nasal septal CPCs. Subsequently, we described the roles of migratory CPCs in cartilage regeneration. Finally, we reviewed the existing studies on CPCs-based cartilage tissue engineering and summarized the strategies for promoting the migration and chondrogenesis of CPCs so as to provide ideas for achieving nasal septal cartilage in situ regeneration.
Collapse
Affiliation(s)
| | | | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; (C.Z.)
| |
Collapse
|
13
|
Rodríguez-Cendal AI, Gómez-Seoane I, de Toro-Santos FJ, Fuentes-Boquete IM, Señarís-Rodríguez J, Díaz-Prado SM. Biomedical Applications of the Biopolymer Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV): Drug Encapsulation and Scaffold Fabrication. Int J Mol Sci 2023; 24:11674. [PMID: 37511432 PMCID: PMC10380382 DOI: 10.3390/ijms241411674] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) is a biodegradable and biocompatible biopolymer that has gained popularity in the field of biomedicine. This review provides an overview of recent advances and potential applications of PHBV, with special emphasis on drug encapsulation and scaffold construction. PHBV has shown to be a versatile platform for drug delivery, offering controlled release, enhanced therapeutic efficacy, and reduced side effects. The encapsulation of various drugs, such as anticancer agents, antibiotics, and anti-inflammatory drugs, in PHBV nanoparticles or microspheres has been extensively investigated, demonstrating enhanced drug stability, prolonged release kinetics, and increased bioavailability. Additionally, PHBV has been used as a scaffold material for tissue engineering applications, such as bone, cartilage, and skin regeneration. The incorporation of PHBV into scaffolds has been shown to improve mechanical properties, biocompatibility, and cellular interactions, making them suitable for tissue engineering constructs. This review highlights the potential of PHBV in drug encapsulation and scaffold fabrication, showing its promising role in advancing biomedical applications.
Collapse
Affiliation(s)
- Ana Isabel Rodríguez-Cendal
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade de A Coruña, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - Iván Gómez-Seoane
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - Francisco Javier de Toro-Santos
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade de A Coruña, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Servicio de Reumatología, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - Isaac Manuel Fuentes-Boquete
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade de A Coruña, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - José Señarís-Rodríguez
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Servicio de Cirugía Ortopédica y Traumatología, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
| | - Silvia María Díaz-Prado
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade de A Coruña, Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), 15008 A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
14
|
Feng K, Yu Y, Chen Z, Wang F, Zhang K, Chen H, Xu J, Kang Q. Injectable hypoxia-preconditioned cartilage progenitor cells-laden GelMA microspheres system for enhanced osteoarthritis treatment. Mater Today Bio 2023; 20:100637. [PMID: 37128287 PMCID: PMC10148185 DOI: 10.1016/j.mtbio.2023.100637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023] Open
Abstract
Osteoarthritis (OA) is the most common age-related degenerative joint disease mainly characterized by the destruction of articular cartilage. Owing to its native avascular property, intrinsic repair of articular cartilage is very limited. Thus, a chondrogenic microenvironment in the joint is essential to the preservation of healthy chondrocytes and OA treatment. Recently, cartilage progenitor cells (CPCs)-based therapy is emerging as a promising strategy to repair degenerated and damaged articular cartilage. In this study, injectable hypoxia-preconditioned three-dimensional (3D) cultured CPCs-laden gelatin methacryloyl (GelMA) microspheres (CGMs) were constructed and characterized. Compared to normoxia-pretreated 3D CPCs and two-dimensional (2D) cultured CPCs, hypoxia-preconditioned 3D cultured CPCs exhibited enhanced cartilage extracellular matrix (ECM) secretion and greater chondrogenic ability. In addition, hypoxia-preconditioned 3D cultured CPCs more effectively maintained cartilage matrix metabolism balance and attenuated articular cartilage degeneration in subacute and chronic rat OA models. Mechanistically, our results demonstrated hypoxia-preconditioned 3D cultured CPCs exerted chondro-protective effects by inhibiting inflammation and oxidative stress via NRF2/HO-1 pathway in vitro and in vivo. Together, through the 3D culture of CPCs using GelMA microspheres (GMs) under hypoxia environment, our results proposed an efficient articular cartilage regeneration strategy for OA treatment and could provide inspiration for other stem cells-based therapies.
Collapse
Affiliation(s)
- Kai Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yifan Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhengsheng Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Kunqi Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongfang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
15
|
Vinod E, Parasuraman G, Livingston A, Amirtham SM, Rebekah G, Lisha JJ, Daniel AJ, Sathishkumar S. Comparison of methods for the isolation and culture of Migratory chondroprogenitors from Human articular cartilage. Connect Tissue Res 2023:1-11. [PMID: 37092666 DOI: 10.1080/03008207.2023.2202266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
PURPOSE Resident articular stem cells isolated using a migratory assay called Migratory Chondroprogenitors (MCPs) have emerged as a promising cellular therapeutic for the treatment of cartilage pathologies. In-vivo studies using MCPs report their superiority over bone-marrow mesenchymal stem cells and chondrocytes for treating chondral defects. However, there is no consensus on their isolation protocol. This study aimed to compare four reported isolation methods of MCPs and identify the optimal and feasible protocol for future translational work. METHODS Human MCPs isolated from osteoarthritic cartilage (n = 3) were divided into four groups: a) MCP1: 8-15 mm cartilage explants, b) MCP2: 8-10 mm explants digested in 0.1% collagenase for 2 hrs. and cultured c) MCP3: 1 mm cartilage explants and d) MCP 4: 25 mm explants with a X tear, 7-day culture, and trypsinization to release migrated cells. The MCPs were subjected to the following analysis: growth kinetics, surface marker expression, mRNA gene expression for markers of chondrogenesis and hypertrophy, and trilineage differentiation. RESULTS MCPs isolated via the four methods showed similar surface marker profiles, chondrogenic (SOX-9, ACAN, COL2A1) and hypertrophic (COL1, RUNX2) gene expression. The migration time for the MCP3 group was the longest. The MCP1, MCP2, and MCP4 groups produced MCPs with comparable cellular expansion feasibility. CONCLUSIONS MCPs can be preferably isolated by the any of the three above methods based on the investigator's discretion. In the case of small cartilage samples similar to the MCP3 group, the isolation of MCP is plausible, keeping in mind the additional time required.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India
- Centre for Stem Cell Research, A unit of InStem, Bengaluru, India
| | | | - Abel Livingston
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | | | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, India
| | - J Jeya Lisha
- Department of Physiology, Christian Medical College, Vellore, India
| | - Alfred Job Daniel
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | | |
Collapse
|
16
|
Zhang X, Qi L, Chen X, Lai Y, Liu K, Xue K. Comparative study of alginate and type I collagen as biomaterials for cartilage stem/progenitor cells to construct tissue-engineered cartilage in vivo. Front Bioeng Biotechnol 2023; 10:1057199. [PMID: 36714619 PMCID: PMC9873961 DOI: 10.3389/fbioe.2022.1057199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
With the help of biomaterials, cartilage stem/progenitor cells (CSPCs) derived from cartilage tissue present a promising choice for cartilage regeneration. In our previous study, we investigated whether CSPCs could be ideal seeding cells for cartilage tissue regeneration. Biomaterials are fabricated to accelerate tissue regeneration, providing a suitable environment for cell attachment, proliferation, and differentiation. Among the biomaterials used in cartilage regeneration medicine, alginate and collagen are classified as natural biomaterials and are characterized by high biocompatibility, bioactivity, and non-toxic degradation products. However, it is unclear which material would have a competitive advantage in CSPC-based cartilage regeneration in vivo. In the present study, we employed alginate and type Ⅰ collagen as substrates for CSPCs and chondrocytes, which was made control group, to explore a more suitable biomaterials for CSPCs to fabricate tissue-engineered cartilage, in vivo. Hematoxylin and eosin (HE) staining, Safranin O, immunohistochemical assay, and quantitative real-time polymerase chain reaction (qRT-PCR) were used to evaluate the tissue-engineered cartilage in vivo. Compared with the alginate group, collagen enhanced the expression of cartilage-specific genes, such as ACAN, SOX9, and COLII, more markedly. Furthermore, the marker genes of expression, dedifferentiation, and hypertrophy, COLI and COLX, were downregulated in the collagen group. The results demonstrated that collagen as a substrate was superior to alginate in increasing the accumulation of cartilage-like ECM for CSPCs in vivo. In summary, compared with alginate, collagen hydrogel is an effective biomaterial for CSPC-based cartilage regeneration.
Collapse
Affiliation(s)
- Xiaodie Zhang
- Department of Dermatologic Surgery, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Qi
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - XiaoGang Chen
- Department of Dermatologic Surgery, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongxian Lai
- Department of Dermatologic Surgery, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kai Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, Hainan Western Central Hospital, Shanghai, China
| |
Collapse
|
17
|
Vinod E, Padmaja K, Ramasamy B, Sathishkumar S. Systematic review of articular cartilage derived chondroprogenitors for cartilage repair in animal models. J Orthop 2023; 35:43-53. [PMID: 36387762 PMCID: PMC9647330 DOI: 10.1016/j.jor.2022.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
Purpose of research The potential for cartilage repair using articular cartilage derived chondroprogenitors has recently gained popularity due to promising results from in-vitro and in-vivo studies. Translation of results from in-vitro to a clinical setting requires a sufficient number of animal studies displaying significant positive outcomes. Thus, this systematic review comprehensively discusses the available literature (January 2000-March 2022) on animal models employing chondroprogenitors for cartilage regeneration, highlighting the results and limitations associated with their use.As per Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a web-based search of PubMed and SCOPUS databases was performed for the following terminologies: "chondroprogenitors", "cartilage-progenitors", and "chondrogenic-progenitors", which yielded 528 studies. A total of 12 studies met the standardized inclusion criteria, which included chondroprogenitors derived from hyaline cartilage isolated using fibronectin adhesion assay (FAA) or migratory assay from explant cultures, further analyzing the role of chondroprogenitors using in-vivo animal models. Principal results Analysis revealed that FAA chondroprogenitors demonstrated the ability to attenuate osteoarthritis, repair chondral defects and form stable cartilage in animal models. They displayed better outcomes than bone marrow-derived mesenchymal stem cells but were comparable to chondrocytes. Migratory chondroprogenitors also demonstrated superiority to BM-MSCs in terms of higher chondrogenesis and lower hypertrophy, although a direct comparison to FAA-CPs and other cell types is warranted. Major conclusions Chondroprogenitors exhibit superior properties for chondrogenic repair; however, limited data on animal studies necessitates further studies to optimize their use before clinical translation for neo-cartilage formation.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India
- Centre for Stem Cell Research, (A unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Kawin Padmaja
- Department of Physiology, Christian Medical College, Vellore, India
| | - Boopalan Ramasamy
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | | |
Collapse
|
18
|
Yazdanian M, Alam M, Abbasi K, Rahbar M, Farjood A, Tahmasebi E, Tebyaniyan H, Ranjbar R, Hesam Arefi A. Synthetic materials in craniofacial regenerative medicine: A comprehensive overview. Front Bioeng Biotechnol 2022; 10:987195. [PMID: 36440445 PMCID: PMC9681815 DOI: 10.3389/fbioe.2022.987195] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
The state-of-the-art approach to regenerating different tissues and organs is tissue engineering which includes the three parts of stem cells (SCs), scaffolds, and growth factors. Cellular behaviors such as propagation, differentiation, and assembling the extracellular matrix (ECM) are influenced by the cell's microenvironment. Imitating the cell's natural environment, such as scaffolds, is vital to create appropriate tissue. Craniofacial tissue engineering refers to regenerating tissues found in the brain and the face parts such as bone, muscle, and artery. More biocompatible and biodegradable scaffolds are more commensurate with tissue remodeling and more appropriate for cell culture, signaling, and adhesion. Synthetic materials play significant roles and have become more prevalent in medical applications. They have also been used in different forms for producing a microenvironment as ECM for cells. Synthetic scaffolds may be comprised of polymers, bioceramics, or hybrids of natural/synthetic materials. Synthetic scaffolds have produced ECM-like materials that can properly mimic and regulate the tissue microenvironment's physical, mechanical, chemical, and biological properties, manage adherence of biomolecules and adjust the material's degradability. The present review article is focused on synthetic materials used in craniofacial tissue engineering in recent decades.
Collapse
Affiliation(s)
- Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Alam
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Abbasi
- Department of Prosthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Rahbar
- Department of Restorative Dentistry, School of Dentistry, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amin Farjood
- Orthodontic Department, Dental School, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Arian Hesam Arefi
- Dental Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
19
|
Padmaja K, Amirtham SM, Rebekah G, Sathishkumar S, Vinod E. Supplementation of articular cartilage-derived chondroprogenitors with bone morphogenic protein-9 enhances chondrogenesis without affecting hypertrophy. Biotechnol Lett 2022; 44:1037-1049. [PMID: 35920961 DOI: 10.1007/s10529-022-03280-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/05/2022] [Indexed: 11/02/2022]
Abstract
INTRODUCTION Chondroprogenitors (CPCs) have emerged as a promising cellular therapy for cartilage-related pathologies due to their inherent primed chondrogenic potential. Studies report that the addition of growth factors such as parathyroid hormone (PTH) and Bone Morphogenic Protein (BMP) enhance the chondroinducive potential in chondrocytes and mesenchymal stem cells. This study evaluated if supplementation of the standard culture medium for cell expansion with 1-34 PTH and BMP-9 would enhance the chondrogenic potential of CPCs and reduce their hypertrophic tendency. METHODS Human chondrocytes were isolated from patients undergoing total knee replacement for osteoarthritis (n = 3). Following fibronectin adhesion assay, passage 1 CPCs were divided and further expanded under three culture conditions (a) control, i.e., cells continued under standard culture conditions, (b) 1-34 PTH group, additional intermittent 6 h exposure with 1-34 PTH and (c) BMP-9 group, additional BMP-9 during culture expansion. All the groups were evaluated for population-doubling, cell cycle analysis, surface marker and gene expression for chondrogenesis, hypertrophy, multilineage differentiation and GAG (glycosaminoglycan)/DNA following chondrogenic differentiation. RESULTS Concerning growth kinetics, the BMP-9 group exhibited a significantly lower S-phase and population-doubling when compared to the other two groups. Qualitative analysis for chondrogenic potential (Alcian blue, Safranin O staining and Toluidine blue for GAG) revealed that the BMP-9 group exhibited the highest uptake. The BMP-9 group also showed significantly higher COL2A1 expression than the control group, with no change in the hypertrophy marker expression. CONCLUSION BMP-9 can potentially be used as an additive for CPCs expansion, to enhance their chondrogenic potential without affecting their low hypertrophic tendency. The mitigating effects of 1-34PTH on hypertrophy would benefit further investigation when used in combination with BMP-9 to enhance chondrogenesis whilst reducing hypertrophy.
Collapse
Affiliation(s)
- Kawin Padmaja
- Department of Physiology, Christian Medical College, Vellore, 632002, India
| | | | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, 632002, India
| | | | - Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, 632002, India. .,Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, 632002, India.
| |
Collapse
|
20
|
Roles of Cartilage-Resident Stem/Progenitor Cells in Cartilage Physiology, Development, Repair and Osteoarthritis. Cells 2022; 11:cells11152305. [PMID: 35892602 PMCID: PMC9332847 DOI: 10.3390/cells11152305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease that causes irreversible destruction of articular cartilage for which there is no effective treatment at present. Although articular cartilage lacks intrinsic reparative capacity, numerous studies have confirmed the existence of cartilage-resident stem/progenitor cells (CSPCs) in the superficial zone (SFZ) of articular cartilage. CSPCs are characterized by the expression of mesenchymal stromal cell (MSC)-related surface markers, multilineage differentiation ability, colony formation ability, and migration ability in response to injury. In contrast to MSCs and chondrocytes, CSPCs exhibit extensive proliferative and chondrogenic potential with no signs of hypertrophic differentiation, highlighting them as suitable cell sources for cartilage repair. In this review, we focus on the organizational distribution, markers, cytological features and roles of CSPCs in cartilage development, homeostasis and repair, and the application potential of CSPCs in cartilage repair and OA therapies.
Collapse
|
21
|
Xue K, Zhang S, Ge J, Wang Q, Qi L, Liu K. Integration of Bioglass Into PHBV-Constructed Tissue-Engineered Cartilages to Improve Chondrogenic Properties of Cartilage Progenitor Cells. Front Bioeng Biotechnol 2022; 10:868719. [PMID: 35685093 PMCID: PMC9172278 DOI: 10.3389/fbioe.2022.868719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The Poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) scaffold has proven to be a promising three-dimensional (3D) biodegradable and bioactive scaffold for the growth and proliferation of cartilage progenitor cells (CPCs). The addition of Bioglass into PHBV was reported to increase the bioactivity and mechanical properties of the bioactive materials.Methods: In the current study, the influence of the addition of Bioglass into PHBV 3D porous scaffolds on the characteristics of CPC-based tissue-engineered cartilages in vivo were compared. CPCs were seeded into 3D macroporous PHBV scaffolds and PHBV/10% Bioglass scaffolds. The CPC–scaffold constructs underwent 6 weeks in vitro chondrogenic induction culture and were then transplanted in vivo for another 6 weeks to evaluate the difference between the CPC–PHBV construct and CPC–PHBV/10% Bioglass construct in vivo.Results: Compared with the pure PHBV scaffold, the PHBV/10% Bioglass scaffold has better hydrophilicity and a higher percentage of adhered cells. The CPC–PHBV/10%Bioglass construct produced much more cartilage-like tissues with higher cartilage-relative gene expression and cartilage matrix protein production and better biomechanical performance than the CPC–PHBV construct.Conclusion: The addition of Bioglass into 3D PHBV macroporous scaffolds improves the characteristics of CPC-based tissue-engineered cartilages in vivo.
Collapse
Affiliation(s)
- Ke Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, Hainan Western Central Hospital, Hainan, China
| | - Shuqi Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Ge
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Lin Qi
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- *Correspondence: Lin Qi, ; Kai Liu,
| | - Kai Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Lin Qi, ; Kai Liu,
| |
Collapse
|
22
|
Johnson NN, Amirtham SM, Sandya Rani B, Sathishkumar S, Rebekah G, Vinod E. Assessment of the inherent chondrogenic potential of human articular cartilage-derived chondroprogenitors in pellet culture using a novel whole pellet processing approach. J Orthop 2022; 31:45-51. [PMID: 35368732 PMCID: PMC8967706 DOI: 10.1016/j.jor.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/21/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose Cartilage-derived chondroprogenitors have been reported to possess the biological potential for cartilage repair. However, its inherent chondrogenic potential in pellet culture needs evaluation. In-vitro cartilage regeneration models based on pellet cultures have been employed to evaluate the chondrogenic potential of stem cells. Evaluation of the degree of differentiation routinely involves paraffin embedding, sectioning, and immunohistochemical staining of the pellet. However, since chondrogenic differentiation is commonly non-uniform, processing random sections could lead to inaccurate conclusions. The study aimed at assessing the inherent lineage bias of chondroprogenitors with and without chondrogenic induction, using a novel whole pellet processing technique. Methods Human chondroprogenitors (n=3) were evaluated for MSC markers and processed in pellet cultures either with stromal medium (uninduced) or chondrogenic differentiation medium (induced) for 28 days. The whole pellets and the conventional paraffin-embedded sectioned pellets were subjected to Collagen type II immunostaining and assessed using confocal laser microscopy. The staining intensities of the whole pellet were compared to the paraffin sections and revalidated using qRT-PCR for COL2A1 expression. Results Uninduced and induced pellets displayed Collagen type II in all the layers with comparable fluorescence intensities. COL2A1 expression in both pellets was comparable to confocal results. The study demonstrated that uninduced chondroprogenitors in pellet culture possess promising inherent chondrogenic potential. Confocal imaging of whole pellets displayed different degrees of chondrogenic differentiation in the entire pellet, thus its probable in-vivo behavior. Conclusion The novel approach presented in this study could serve as an efficient in-vitro alternative for understanding translational application for cartilage repair.
Collapse
|
23
|
Hayashi RG, Borghesi J, Mario LC, Rabelo ACS, de Almeida da Anunciação AR, Lima MF, Miglino MA, de Oliveira Favaron P, Carreira ACO. Immunophenotyping of progenitor cells from articular cartilage of New Zealand Rabbits (Oryctolagus cuniculus). Tissue Cell 2022; 75:101742. [DOI: 10.1016/j.tice.2022.101742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/28/2022]
|
24
|
Vinod E, Johnson NN, Kumar S, Amirtham SM, James JV, Livingston A, Rebekah G, Daniel AJ, Ramasamy B, Sathishkumar S. Migratory chondroprogenitors retain superior intrinsic chondrogenic potential for regenerative cartilage repair as compared to human fibronectin derived chondroprogenitors. Sci Rep 2021; 11:23685. [PMID: 34880351 PMCID: PMC8654938 DOI: 10.1038/s41598-021-03082-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Cell-based therapy for articular hyaline cartilage regeneration predominantly involves the use of mesenchymal stem cells and chondrocytes. However, the regenerated repair tissue is suboptimal due to the formation of mixed hyaline and fibrocartilage, resulting in inferior long-term functional outcomes. Current preclinical research points towards the potential use of cartilage-derived chondroprogenitors as a viable option for cartilage healing. Fibronectin adhesion assay-derived chondroprogenitors (FAA-CP) and migratory chondroprogenitors (MCP) exhibit features suitable for neocartilage formation but are isolated using distinct protocols. In order to assess superiority between the two cell groups, this study was the first attempt to compare human FAA-CPs with MCPs in normoxic and hypoxic culture conditions, investigating their growth characteristics, surface marker profile and trilineage potency. Their chondrogenic potential was assessed using mRNA expression for markers of chondrogenesis and hypertrophy, glycosaminoglycan content (GAG), and histological staining. MCPs displayed lower levels of hypertrophy markers (RUNX2 and COL1A1), with normoxia-MCP exhibiting significantly higher levels of chondrogenic markers (Aggrecan and COL2A1/COL1A1 ratio), thus showing superior potential towards cartilage repair. Upon chondrogenic induction, normoxia-MCPs also showed significantly higher levels of GAG/DNA with stronger staining. Focused research using MCPs is required as they can be suitable contenders for the generation of hyaline-like repair tissue.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India. .,Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India.
| | | | - Sanjay Kumar
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | | | - Jithu Varghese James
- Department of Diabetes, School of Life Course Sciences, King's College London, London, UK
| | - Abel Livingston
- Department of Orthopaedics, Christian Medical College and Hospital, Vellore, India
| | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, India
| | - Alfred Job Daniel
- Department of Orthopaedics, Christian Medical College and Hospital, Vellore, India
| | - Boopalan Ramasamy
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia. .,Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia.
| | | |
Collapse
|
25
|
Vinod E, Parameswaran R, Ramasamy B, Kachroo U. Pondering the Potential of Hyaline Cartilage-Derived Chondroprogenitors for Tissue Regeneration: A Systematic Review. Cartilage 2021; 13:34S-52S. [PMID: 32840123 PMCID: PMC8804774 DOI: 10.1177/1947603520951631] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Chondroprogenitors have recently gained prominence due to promising results seen in in vitro and animal studies as a potential contender in cell-based therapy for cartilage repair. Lack of consensus regarding nomenclature, isolation techniques, and expansion protocols create substantial limitations for translational research, especially given the absence of distinct markers of identification. The objective of this systematic review was to identify and collate information pertaining to hyaline cartilage-derived chondroprogenitors, with regard to their isolation, culture, and outcome measures. DESIGN As per Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a web-based search of Scopus and PubMed databases was performed from January 2000 to May 2020, which yielded 509 studies. A total of 65 studies were identified that met the standardized inclusion criteria which comprised of, but was not limited to, progenitors derived from fibronectin adhesion, migrated subpopulation from explant cultures, and single-cell sorting. RESULT Literature search revealed that progenitors demonstrated inherent chondrogenesis and minimal tendency for hypertrophy. Multiple sources also demonstrated significantly better outcomes that bone marrow-derived mesenchymal stem cells and comparable results to chondrocytes. With regard to progenitor subgroups, collated evidence points to better and consistent outcomes with the use of migratory progenitors when compared to fibronectin adhesion assay-derived progenitors, although a direct comparison between the two cell populations is warranted. CONCLUSION Since chondroprogenitors exhibit favorable properties for cartilage repair, efficient characterization of progenitors is imperative, to complete their phenotypic profile, so as to optimize their use in translational research for neocartilage formation.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian
Medical College, Vellore, India,Centre for Stem Cell Research (a unit of
InStem, Bengaluru), Christian Medical College, Vellore, India,Elizabeth Vinod, Department of Physiology,
Christian Medical College, Vellore 632002, Tamil Nadu, India.
| | | | - Boopalan Ramasamy
- Department of Orthopaedics, Royal Darwin
Hospital, Tiwi, Northern Territory, Australia
| | - Upasana Kachroo
- Department of Physiology, Christian
Medical College, Vellore, India
| |
Collapse
|
26
|
Xue K, Jiang Y, Zhang X, Wu J, Qi L, Liu K. Hypoxic ADSCs-derived EVs promote the proliferation and chondrogenic differentiation of cartilage stem/progenitor cells. Adipocyte 2021; 10:322-337. [PMID: 34224296 PMCID: PMC8259721 DOI: 10.1080/21623945.2021.1945210] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Cartilage tissue engineering is a promising option for repairing cartilage defects, although harvesting a large number of seeding cells remains a major challenge. Cartilage stem/progenitor cells (CSPCs) seem to be a promising cell source. Hypoxic extracellular vesicles (EVs) may play a major role in cell-cell and tissue-tissue communication. In the current study, we aimed to evaluate the effect of hypoxic adipose-derived stem cells (ADSCs)-derived EVs on CSPCs proliferation and differentiation. The characteristics of ADSCs-derived EVs were identified, and proliferation, migration, and cartilage-related gene expression of CSPCs were measured with or without the presence of hypoxic ADSCs-derived EVs. SEM, histological staining, biochemical and biomechanical analysis was performed to evaluate the effect of hypoxic ADSCs-derived EVs on CSPCs in alginate hydrogel culture. The results indicated that the majority of ADSC-derived EVs exhibited a round-shaped or cup-shaped morphology with a diameter of 40-1000 nm and expressed CD9, CD63, and CD81. CSPCs migration and proliferation were enhanced by hypoxic ADSCs-derived EVs, which also increased the expression of cartilage-related genes. The hypoxic ADSCs-derived EVs induce CSPCs to produce significantly more cartilage matrix and proteoglycan. In conclusion, hypoxic ADSCs-derived EVs improved the proliferation and chondrogenic differentiation of CSPCs for cartilage tissue engineering.
Collapse
Affiliation(s)
- Ke Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Yongkang Jiang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Xiaodie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Jun Wu
- Department of Orthopedics, The First People’s Hospital of Changzhou, Jiangsu Changzhou, China
| | - Lin Qi
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Kai Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| |
Collapse
|
27
|
Peláez P, Damiá E, Torres-Torrillas M, Chicharro D, Cuervo B, Miguel L, del Romero A, Carrillo JM, Sopena JJ, Rubio M. Cell and Cell Free Therapies in Osteoarthritis. Biomedicines 2021; 9:1726. [PMID: 34829953 PMCID: PMC8615373 DOI: 10.3390/biomedicines9111726] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is the most common articular disease in adults and has a current prevalence of 12% in the population over 65 years old. This chronic disease causes damage to articular cartilage and synovial joints, causing pain and leading to a negative impact on patients' function, decreasing quality of life. There are many limitations regarding OA conventional therapies-pharmacological therapy can cause gastrointestinal, renal, and cardiac adverse effects, and some of them could even be a threat to life. On the other hand, surgical options, such as microfracture, have been used for the last 20 years, but hyaline cartilage has a limited regeneration capacity. In recent years, the interest in new therapies, such as cell-based and cell-free therapies, has been considerably increasing. The purpose of this review is to describe and compare bioregenerative therapies' efficacy for OA, with particular emphasis on the use of mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP). In OA, these therapies might be an alternative and less invasive treatment than surgery, and a more effective option than conventional therapies.
Collapse
Affiliation(s)
- Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Elena Damiá
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Belén Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Ayla del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Jose Maria Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Joaquín J. Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Mónica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| |
Collapse
|
28
|
Li M, Ning J, Wang J, Yan Q, Zhao K, Jia X. SETD7 regulates chondrocyte differentiation and glycolysis via the Hippo signaling pathway and HIF‑1α. Int J Mol Med 2021; 48:210. [PMID: 34617577 PMCID: PMC8510680 DOI: 10.3892/ijmm.2021.5043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Chondrocytes are well adapted to hypoxia and produce more functional extracellular matrix in low oxygen environments in vitro. In our previous study, methyltransferase SET domain containing (SETD)7 regulated chondrocyte activity in hypoxic conditions. However, the precise association between SETD7 and chondrocyte differentiation under low oxygen partial pressure remains unclear. The association between SETD7 and chondrocyte differentiation was studied by silencing SETD7 in chondrocytes in vitro. The results showed that the silencing of SETD7 in ATDC5 cells inhibited the Hippo signaling pathway, decreased Yes-associated protein (YAP) phosphorylation and increased the levels of YAP and hypoxia inducible factor-1α (HIF-1α) in the nucleus. YAP combined with HIF-1α to form a complex that promoted the expression of genes involved in chondrogenic differentiation and the glycolytic pathway. Thus, SETD7 inhibited chondrocyte differentiation and glycolysis via the Hippo signaling pathway. The present study demonstrated that SETD7 was a potential molecular target that maintained the chondrocyte phenotype during cartilage tissue engineering and cartilage-associated disease.
Collapse
Affiliation(s)
- Maoquan Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Jinqiu Ning
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Jiwei Wang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Qiqian Yan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Ke Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xiaoshi Jia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
29
|
Chen W, Li Y, Huang Y, Dai Y, Xi T, Zhou Z, Liu H. Quercetin modified electrospun PHBV fibrous scaffold enhances cartilage regeneration. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:92. [PMID: 34374884 PMCID: PMC8354921 DOI: 10.1007/s10856-021-06565-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
It suggests that the poly (3-hydroxybutyric acid-co-3-hydroxyvaleric acid) (PHBV) scaffold can be used for cartilage tissue engineering, but PHBV is short of bioactivity that is required for cartilage regeneration. To fabricate a bioactive cartilage tissue engineering scaffold that promotes cartilage regeneration, quercetin (QUE) modified PHBV (PHBV-g-QUE) fibrous scaffolds were prepared by a two-step surface modification method. The PHBV-g-QUE fibrous scaffold facilitates the growth of chondrocytes and maintains chondrocytic phenotype resulting from the upregulation of SOX9, COL II, and ACAN. The PHBV-g-QUE fibrous scaffold inhibited apoptosis of chondrocyte and reduced oxidative stress of chondrocytes by regulating the transcription of related genes. Following PHBV-g-QUE fibrous scaffolds and PHBV fibrous scaffolds with adhered chondrocytes were implanted into nude mice for 4 weeks, it demonstrated that PHBV-g-QUE fibrous scaffolds significantly promoted cartilage regeneration compared with the PHBV fibrous scaffolds. Hence, it suggests that the PHBV-g-QUE fibrous scaffold can be potentially applied in the clinical treatment of cartilage defects in the future.
Collapse
Affiliation(s)
- Wei Chen
- College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Yongsheng Li
- College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Yuting Huang
- College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Yao Dai
- College of Materials Science and Engineering, Hunan University, Changsha, 410082, China.
| | - Tingfei Xi
- Shenzhen Institute, Peking University, Shenzhen, 518057, China
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha, 410082, China
| | - Hairong Liu
- College of Materials Science and Engineering, Hunan University, Changsha, 410082, China.
- Hunan Province Key Laboratory for Spray Deposition Technology and Application, Hunan University, Changsha, 410082, China.
| |
Collapse
|
30
|
Vinod E, Amirtham SM, Kachroo U, Goyal A, Ozbey O, James JV, Sathishkumar S, Ramasamy B. Articular chondroprogenitors in platelet rich plasma for treatment of osteoarthritis and osteochondral defects in a rabbit knee model. Knee 2021; 30:51-62. [PMID: 33857741 DOI: 10.1016/j.knee.2021.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/22/2020] [Accepted: 03/18/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Articular chondroprogenitors are a promising contender for cartilage repair due to their inherent nature which stands primed for chondrogenesis and minimal hypertrophic preponderance. Platelet rich plasma (PRP) has been extensively used for treating cartilage defects and osteoarthritis (OA), due to its chondro-inductive properties and abundant pool of growth factors. The aim of this study was to assess the efficacy of chondroprogenitors injected with PRP versus PRP alone in the healing of experimentally created early OA and osteochondral defects (OCD) in a rabbit model. METHODS Adult New Zealand White male rabbits were used for cell and PRP isolation. Chondroprogenitors were isolated by fibronectin adhesion assay, labelled with iron oxide, characterized for surface markers, differential potential and expanded. PRP was isolated by double spin centrifugation using a TriCell kit. Study groups included (a) Monosodium iodoacetate induced early OA and (b) critical OCD. Following intervention (test arm: PRP+ chondroprogenitors and control arm: PRP), assessment was performed at 6- and 12-weeks which included histopathological examination and scoring (OARSI and Modified Wakitani score), immunohistochemistry analysis (Collagen type II and X) and synovial fluid S100A12 levels. RESULTS AND CONCLUSION Comparable, evident healing was noticed in both test and control arms when the OA group samples were assessed at both time points. In the OCD group, PRP alone exhibited significantly better results than the test arm, although repair was notable in both interventions. Further evaluation of chondroprogenitors is required to assess their role as a standalone therapy and in combination with PRP to further cartilage regeneration.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore 632002, India; Centre for Stem Cell Research, Christian Medical College, Vellore 632002, India.
| | | | - Upasana Kachroo
- Department of Physiology, Christian Medical College, Vellore 632002, India
| | - Anjali Goyal
- Department of Pathology, Smt NHL Municipal Medical College, Ahmedabad 380006, India.
| | - Ozlem Ozbey
- Department of Histology and Embryology Campus, School of Medicine, Akdeniz University, Antalya 07070, Turkey
| | | | | | - Boopalan Ramasamy
- Department of Orthopaedics, Royal Darwin Hospital, 105 Rocklands Drive, Tiwi, NT 0810, Australia.
| |
Collapse
|
31
|
Zheng J, Lin Y, Tang F, Guo H, Yan L, Hu S, Wu H. Promotive Role of CircATRNL1 on Chondrogenic Differentiation of BMSCs Mediated by miR-338-3p. Arch Med Res 2021; 52:514-522. [PMID: 33610389 DOI: 10.1016/j.arcmed.2021.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 01/05/2021] [Accepted: 02/04/2021] [Indexed: 01/22/2023]
Abstract
AIM Bone marrow mesenchymal stem cells (BMSCs) are ideal seed cells for tissue engineering cartilage construction. However, the underlying mechanism of it has not been illuminate well. In this study, the effects of circATRNL1 (hsa_circ_0020093) on the differentiation of BMSCs into chondrocytes were investigated. METHODS The degrees of chondrogenic differentiation of BMSCs on day 0, 14 and 21 mediums were detected by Alcian blue staining. Expressions of cartilage differentiation related factors SOX9, COL2 and Aggrecan, and circATRNL1 in BMSCs under differentiation were determined by western blot and quantitative real-time polymerase chain reaction (qRT-PCR) as needed. circATRNL1 knockdown or overexpression was performed in BMSCs. Then the viability of BMSCs and cartilage differentiation related factors were separately investigated through MTT assay, qRT-PCR, and western blot. Target gene of circATRNL1 and binding site were predicted using starbase and validated it by dual luciferase reporter. The effect of circATRNL1 and its target gene on chondrogenic differentiation of BMSCs was assessed using Alcian blue staining further. RESULTS The degrees of chondrogenic differentiation of BMSCs were increased with time. Expressions of SOX9, COL2 and Aggrecan as well as circATRNL1 were enhanced during chondrogenic differentiation. Furthermore, overexpression of circATRNL1 enhanced BMSCs proliferation, SOX9, COL2 and Aggrecan expressions and the degree of chondrogenic differentiation of BMSCs. Further research showed that circATRNL1 targeted miR-338-3p. MiR-338-3p inhibited differentiation of BMSCs into cartilage but overexpression of circATRNL1 reversed it. CONCLUSION CircATRNL1 is beneficial to BMSCs differentiation into cartilage by regulating miR-338-3p, which may be a new mechanism of action in the treatment of cartilage repair.
Collapse
Affiliation(s)
- Jianzhang Zheng
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China
| | - Yunshuo Lin
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China
| | - Faqiang Tang
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China
| | - Huiling Guo
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China
| | - Laipeng Yan
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China
| | - Shiping Hu
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China
| | - Hong Wu
- Shengli Clinical Medical College of Fujian Medical University, Department of Orthopaedics, Fujian Provincial Hospital, Fuzhou, China.
| |
Collapse
|
32
|
Schmidt S, Abinzano F, Mensinga A, Teßmar J, Groll J, Malda J, Levato R, Blunk T. Differential Production of Cartilage ECM in 3D Agarose Constructs by Equine Articular Cartilage Progenitor Cells and Mesenchymal Stromal Cells. Int J Mol Sci 2020; 21:ijms21197071. [PMID: 32992847 PMCID: PMC7582568 DOI: 10.3390/ijms21197071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Identification of articular cartilage progenitor cells (ACPCs) has opened up new opportunities for cartilage repair. These cells may be used as alternatives for or in combination with mesenchymal stromal cells (MSCs) in cartilage engineering. However, their potential needs to be further investigated, since only a few studies have compared ACPCs and MSCs when cultured in hydrogels. Therefore, in this study, we compared chondrogenic differentiation of equine ACPCs and MSCs in agarose constructs as monocultures and as zonally layered co-cultures under both normoxic and hypoxic conditions. ACPCs and MSCs exhibited distinctly differential production of the cartilaginous extracellular matrix (ECM). For ACPC constructs, markedly higher glycosaminoglycan (GAG) contents were determined by histological and quantitative biochemical evaluation, both in normoxia and hypoxia. Differential GAG production was also reflected in layered co-culture constructs. For both cell types, similar staining for type II collagen was detected. However, distinctly weaker staining for undesired type I collagen was observed in the ACPC constructs. For ACPCs, only very low alkaline phosphatase (ALP) activity, a marker of terminal differentiation, was determined, in stark contrast to what was found for MSCs. This study underscores the potential of ACPCs as a promising cell source for cartilage engineering.
Collapse
Affiliation(s)
- Stefanie Schmidt
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany;
| | - Florencia Abinzano
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
| | - Anneloes Mensinga
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
| | - Jörg Teßmar
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany; (J.T.); (J.G.)
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany; (J.T.); (J.G.)
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
- Correspondence: (R.L.); (T.B.)
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany;
- Correspondence: (R.L.); (T.B.)
| |
Collapse
|
33
|
Nahanmoghadam A, Asemani M, Goodarzi V, Ebrahimi-Barough S. Design and fabrication of bone tissue scaffolds based on PCL/PHBV containing hydroxyapatite nanoparticles: dual-leaching technique. J Biomed Mater Res A 2020; 109:981-993. [PMID: 33448637 DOI: 10.1002/jbm.a.37087] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/09/2020] [Accepted: 08/14/2020] [Indexed: 01/11/2023]
Abstract
Scaffolds are the important part of the tissue-engineering field that are made from different biomaterials using various techniques. In this study, new scaffold based on polycaprolactone (PCL) and poly (hydroxybutyrate-co-hydroxyvalerate) (PHBV) containing hydroxyapatite nanopraticles (n-HA) were fabricated using the dual-leaching technique (DLT). Morphology, porosity, degradation rate, Fourier transfer infrared ray (FTIR) spectra, surface, and mechanical properties as well as capacity of cell binding and cell proliferation on the constructed scaffolds were evaluated. FTIR analysis showed that n-HA particles have some interest interactions with polymeric chains. The best 3D-structure was seen in PCL70PHBV30 scaffold using the scanning electron microscopy (SEM) and its structure improved in the presence of 3, 5 wt% of n-HA. Results of energy dispersive x-ray analysis (EDXA, map of Ca) showed that the nanoparticles have the uniform distribution within the fabricated scaffolds. Porosity analysis showed that the particulate salt leaching technique is a successful approach to building a 3D structure. Increasing of PHBV content and n-HA up to 3 and 5 wt% in the PCL matrix led to increase porosity in all samples. Mechanical properties analysis showed that values of compression modulus and strength are decreased with addition of PHBV and HA nanoparticles. These results were directly in line with the results of morphology and porosity. Cell culture experiments demonstrated that the PCL/PHBV/nHA nanocomposite scaffold has a better tendency of proliferation to cells than that of the pure PCL/PHBV scaffold. All of these results suggest promising potentials of the developed PCL/PHBV/nHA scaffolds in this study desire for bone tissue engineering.
Collapse
Affiliation(s)
- Amir Nahanmoghadam
- Department of Chemical Engineering, Faculty of Engineering, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Maryam Asemani
- Department of Chemical Engineering, Faculty of Engineering, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Vahabodin Goodarzi
- Applied biotechnology research center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Rahmani Del Bakhshayesh A, Babaie S, Tayefi Nasrabadi H, Asadi N, Akbarzadeh A, Abedelahi A. An overview of various treatment strategies, especially tissue engineering for damaged articular cartilage. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:1089-1104. [DOI: 10.1080/21691401.2020.1809439] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Azizeh Rahmani Del Bakhshayesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soraya Babaie
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Sha'ban M, Ahmad Radzi MA. Scaffolds for Cartilage Regeneration: To Use or Not to Use? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1249:97-114. [PMID: 32602093 DOI: 10.1007/978-981-15-3258-0_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Joint cartilage has been a significant focus on the field of tissue engineering and regenerative medicine (TERM) since its inception in the 1980s. Represented by only one cell type, cartilage has been a simple tissue that is thought to be straightforward to deal with. After three decades, engineering cartilage has proven to be anything but easy. With the demographic shift in the distribution of world population towards ageing, it is expected that there is a growing need for more effective options for joint restoration and repair. Despite the increasing understanding of the factors governing cartilage development, there is still a lot to do to bridge the gap from bench to bedside. Dedicated methods to regenerate reliable articular cartilage that would be equivalent to the original tissue are still lacking. The use of cells, scaffolds and signalling factors has always been central to the TERM. However, without denying the importance of cells and signalling factors, the question posed in this chapter is whether the answer would come from the methods to use or not to use scaffold for cartilage TERM. This paper presents some efforts in TERM area and proposes a solution that will transpire from the ongoing attempts to understand certain aspects of cartilage development, degeneration and regeneration. While an ideal formulation for cartilage regeneration has yet to be resolved, it is felt that scaffold is still needed for cartilage TERM for years to come.
Collapse
Affiliation(s)
- Munirah Sha'ban
- Department of Physical Rehabilitation Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia.
| | - Muhammad Aa'zamuddin Ahmad Radzi
- Department of Biomedical Science, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| |
Collapse
|
36
|
Pathomechanisms of Posttraumatic Osteoarthritis: Chondrocyte Behavior and Fate in a Precarious Environment. Int J Mol Sci 2020; 21:ijms21051560. [PMID: 32106481 PMCID: PMC7084733 DOI: 10.3390/ijms21051560] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic injuries of the knee joint result in a wide variety of pathomechanisms, which contribute to the development of so-called posttraumatic osteoarthritis (PTOA). These pathogenetic processes include oxidative stress, excessive expression of catabolic enzymes, release of damage-associated molecular patterns (DAMPs), and synovial inflammation. The present review focuses on the underlying pathomechanisms of PTOA and in particular the behavior and fate of the surviving chondrocytes, comprising chondrocyte metabolism, regulated cell death, and phenotypical changes comprising hypertrophy and senescence. Moreover, possible therapeutic strategies, such as chondroanabolic stimulation, anti-oxidative and anti-inflammatory treatment, as well as novel therapeutic targets are discussed.
Collapse
|
37
|
Voinova V, Bonartseva G, Bonartsev A. Effect of poly(3-hydroxyalkanoates) as natural polymers on mesenchymal stem cells. World J Stem Cells 2019; 11:764-786. [PMID: 31692924 PMCID: PMC6828591 DOI: 10.4252/wjsc.v11.i10.764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/17/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are stromal multipotent stem cells that can differentiate into multiple cell types, including fibroblasts, osteoblasts, chondrocytes, adipocytes, and myoblasts, thus allowing them to contribute to the regeneration of various tissues, especially bone tissue. MSCs are now considered one of the most promising cell types in the field of tissue engineering. Traditional petri dish-based culture of MSCs generate heterogeneity, which leads to inconsistent efficacy of MSC applications. Biodegradable and biocompatible polymers, poly(3-hydroxyalkanoates) (PHAs), are actively used for the manufacture of scaffolds that serve as carriers for MSC growth. The growth and differentiation of MSCs grown on PHA scaffolds depend on the physicochemical properties of the polymers, the 3D and surface microstructure of the scaffolds, and the biological activity of PHAs, which was discovered in a series of investigations. The mechanisms of the biological activity of PHAs in relation to MSCs remain insufficiently studied. We suggest that this effect on MSCs could be associated with the natural properties of bacteria-derived PHAs, especially the most widespread representative poly(3-hydroxybutyrate) (PHB). This biopolymer is present in the bacteria of mammalian microbiota, whereas endogenous poly(3-hydroxybutyrate) is found in mammalian tissues. The possible association of PHA effects on MSCs with various biological functions of poly(3-hydroxybutyrate) in bacteria and eukaryotes, including in humans, is discussed in this paper.
Collapse
Affiliation(s)
- Vera Voinova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119234, Russia
| | - Garina Bonartseva
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow 119071, Russia
| | - Anton Bonartsev
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow 119234, Russia
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow 119071, Russia.
| |
Collapse
|